Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Virol ; 97(4): e0009523, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37014223

ABSTRACT

Many RING domain E3 ubiquitin ligases play critical roles in fine-tuning the innate immune response, yet little is known about their regulatory role in flavivirus-induced innate immunity. In previous studies, we found that the suppressor of cytokine signaling 1 (SOCS1) protein mainly undergoes lysine 48 (K48)-linked ubiquitination. However, the E3 ubiquitin ligase that promotes the K48-linked ubiquitination of SOCS1 is unknown. In the present study, we found that RING finger protein 123 (RNF123) binds to the SH2 domain of SOCS1 through its RING domain and facilitates the K48-linked ubiquitination of the K114 and K137 residues of SOCS1. Further studies found that RNF123 promoted the proteasomal degradation of SOCS1 and promoted Toll-like receptor 3 (TLR3)- and interferon (IFN) regulatory factor 7 (IRF7)-mediated type I IFN production during duck Tembusu virus (DTMUV) infection through SOCS1, ultimately inhibiting DTMUV replication. Overall, these findings demonstrate a novel mechanism by which RNF123 regulates type I IFN signaling during DTMUV infection by targeting SOCS1 degradation. IMPORTANCE In recent years, posttranslational modification (PTM) has gradually become a research hot spot in the field of innate immunity regulation, and ubiquitination is one of the critical PTMs. DTMUV has seriously endangered the development of the waterfowl industry in Southeast Asian countries since its outbreak in 2009. Previous studies have shown that SOCS1 is modified by K48-linked ubiquitination during DTMUV infection, but E3 ubiquitin ligase catalyzing the ubiquitination of SOCS1 has not been reported. Here, we identify for the first time that RNF123 acts as an E3 ubiquitin ligase that regulates TLR3- and IRF7-induced type I IFN signaling during DTMUV infection by targeting the K48-linked ubiquitination of the K114 and K137 residues of SOCS1 and the proteasomal degradation of SOCS1.


Subject(s)
Flavivirus Infections , Flavivirus , Interferon Type I , Suppressor of Cytokine Signaling 1 Protein , Animals , Ducks , Flavivirus/physiology , Immunity, Innate/immunology , Interferon Type I/immunology , Toll-Like Receptor 3/metabolism , Ubiquitin-Protein Ligases/immunology , Ubiquitination , Suppressor of Cytokine Signaling 1 Protein/immunology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Protein Binding , Protein Domains/immunology , Virus Replication , HEK293 Cells , Embryo, Mammalian , Humans
2.
BMC Pulm Med ; 22(1): 145, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35428280

ABSTRACT

BACKGROUND: In this study, we established a chronic obstructive pulmonary disease (COPD) model by stimulating mice with cigarette smoke, and observed the effects of dendritic cells (DCs) overexpressing SOCS1 on Th17, Treg and other related cytokines in peripheral blood, bronchoalveolar lavage fluid and lung tissues of COPD mice. METHODS: After successfully transfecting DCs with overexpressing SOCS1 (DC-SOCS1), the mice were injected with DC-SOCS1 (1 × 106), DC-SOCS1 (2 × 106) and immature DCs (1 × 106) via tail vein on days 1 and 7 of COPD fumigation modeling. After day 28 of modeling, the peripheral blood, BALF and lung tissue samples were extracted from the mice, and the changes of DCs, Th17 and Treg cells and related cytokines were detected by immunohistochemistry, immunofluorescence, HE staining, flow cytometry and ELISA. RESULTS: The results showed that DC-SOCS1 was able to reduce the secretion of pro-inflammatory factors and increase the anti-inflammatory factors in the COPD mice, and the effect of high concentration (2 × 106 DC-SOCS1) was better than low concentration (1 × 106 DC-SOCS1). Moreover, the intervention effect was significant on day 1 compared with day 7. In the mice injected with DC-SOCS1, the expression of CD83, IL-4, Foxp3, and CCR6 was increased on day 1 than those on day 7, while IL-17 and IFN-γ was decreased. CONCLUSIONS: Intervention of COPD mice with high concentrations of DCs-SOCS1 reduced pro-inflammatory factor secretion and attenuated the inflammatory response in COPD. Trial registration Not applicable.


Subject(s)
Cytokines , Dendritic Cells , Pulmonary Disease, Chronic Obstructive , Suppressor of Cytokine Signaling 1 Protein , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Cytokines/immunology , Dendritic Cells/immunology , Mice , Pulmonary Disease, Chronic Obstructive/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
3.
Sci Immunol ; 6(66): eabe8219, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34860579

ABSTRACT

Although CD8+ T cells undergo autonomous clonal proliferation after antigen stimulation in vivo, the expansion of activated CD4+ T cells is limited by intrinsic factors that are poorly characterized. Using genome-wide CRISPR-Cas9 screens and an in vivo system modeling of antigen-experienced CD4+ T cell recruitment and proliferation during a localized immune response, we identified suppressor of cytokine signaling 1 (SOCS1) as a major nonredundant checkpoint imposing a brake on CD4+ T cell proliferation. Using anti­interleukin-2 receptor (IL-2R) blocking antibodies, interferon-γ receptor (IFN-γR) knockout mice, and transcriptomic analysis, we show that SOCS1 is a critical node integrating both IL-2 and IFN-γ signals to block multiple downstream signaling pathways abrogating CD4+ T helper 1 (TH1) cell response. Inactivation of SOCS1 in both murine and human CD4+ T cell antitumor adoptive therapies restored intratumor accumulation, proliferation/survival, persistence, and polyfunctionality and promoted rejection of established tumors. However, in CD8+ T cells, SOCS1 deletion did not affect the proliferation but rather improved survival and effector functions, which allowed for optimal therapeutic outcome when associated with SOCS1 inactivation in CD4+ T cells. Together, these findings identify SOCS1 as a major intracellular negative checkpoint of adoptive T cell response, opening new possibilities to optimize CAR-T cell therapy composition and efficacy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Th1 Cells/immunology , Animals , Female , Male , Mice , Mice, Knockout , Mice, Transgenic
4.
PLoS Pathog ; 17(3): e1009387, 2021 03.
Article in English | MEDLINE | ID: mdl-33690673

ABSTRACT

The skin innate immune response to methicillin-resistant Staphylococcus aureus (MRSA) culminates in the formation of an abscess to prevent bacterial spread and tissue damage. Pathogen recognition receptors (PRRs) dictate the balance between microbial control and injury. Therefore, intracellular brakes are of fundamental importance to tune the appropriate host defense while inducing resolution. The intracellular inhibitor suppressor of cytokine signaling 1 (SOCS-1), a known JAK/STAT inhibitor, prevents the expression and actions of PRR adaptors and downstream effectors. Whether SOCS-1 is a molecular component of skin host defense remains to be determined. We hypothesized that SOCS-1 decreases type I interferon production and IFNAR-mediated antimicrobial effector functions, limiting the inflammatory response during skin infection. Our data show that MRSA skin infection enhances SOCS-1 expression, and both SOCS-1 inhibitor peptide-treated and myeloid-specific SOCS-1 deficient mice display decreased lesion size, bacterial loads, and increased abscess thickness when compared to wild-type mice treated with the scrambled peptide control. SOCS-1 deletion/inhibition increases phagocytosis and bacterial killing, dependent on nitric oxide release. SOCS-1 inhibition also increases the levels of type I and type II interferon levels in vivo. IFNAR deletion and antibody blockage abolished the beneficial effects of SOCS-1 inhibition in vivo. Notably, we unveiled that hyperglycemia triggers aberrant SOCS-1 expression that correlates with decreased overall IFN signatures in the infected skin. SOCS-1 inhibition restores skin host defense in the highly susceptible hyperglycemic mice. Overall, these data demonstrate a role for SOCS-1-mediated type I interferon actions in host defense and inflammation during MRSA skin infection.


Subject(s)
Interferon Type I/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Skin Infections/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Animals , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Skin/immunology , Skin/microbiology , Staphylococcal Skin Infections/microbiology , Suppressor of Cytokine Signaling 1 Protein/metabolism
5.
Int Immunopharmacol ; 95: 107495, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33684877

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is characterized by chondrocyte injury and dysfunction, such as excessive apoptosis, inflammatory response and extracellular matrix (ECM) degradation. Circular RNA (circRNA) deregulation is reported to be involved in OA. Our study aimed to explore the role of circ_0134111 in OA. METHODS: Human chondrocytes were treated with interleukin-1ß (IL-1ß) to mimic OA cell model. The expression of circ_0134111, miR-515-5p and suppressor of cytokine signaling 1 (SOCS1) mRNA was measured by real-time quantitative polymerase chain reaction (RT-qPCR), and the protein levels of SOCS1 and apoptosis-/inflammation-/ECM-related markers were determined by western blot. Cell proliferation and cell apoptosis were assessed using cell counting kit-8 (CCK-8) and flow cytometry assay, respectively. For mechanism analysis, the predicted interaction between miR-515-5p and circ_0134111 or SOCS1 was verified by dual-luciferase reporter assay, pull-down assay and RNA immunoprecipitation (RIP) assay. Rescue experiments were performed to explore the interplay between miR-515-5p and circ_0134111 or SOCS1. RESULTS: Circ_0134111 was overexpressed in OA cartilage tissues and IL-1ß-induced chondrocytes. IL-1ß-induced chondrocyte apoptosis, inflammatory responses and ECM degradation were alleviated by circ_0134111 knockdown or miR-515-5p restoration. Circ_0134111 acted as miR-515-5p sponge to regulate miR-515-5p expression, and miR-515-5p deficiency reversed the effects of circ_0134111 knockdown in IL-1ß-induced chondrocytes. MiR-515-5p directly bound to SOCS1, and circ_0134111 decoyed miR-515-5p to increase SOCS1 level. MiR-515-5p restoration alleviated IL-1ß-induced chondrocyte apoptosis, inflammatory responses and ECM degradation, While SOCS1 overexpression partly abolished these effects. CONCLUSION: Circ_0134111 knockdown alleviated apoptosis, inflammatory responses and ECM degradation in OA cell model by mediating the miR-515-5p-SOCS1 network, hinting that circ_0134111 was involved in OA progression.


Subject(s)
Interleukin-1beta/immunology , MicroRNAs/immunology , Osteoarthritis/immunology , RNA, Circular , Suppressor of Cytokine Signaling 1 Protein/immunology , Apoptosis/genetics , Apoptosis/immunology , Cartilage/immunology , Chondrocytes/immunology , Extracellular Matrix/immunology , Female , Humans , Male , Middle Aged
6.
Front Immunol ; 11: 582102, 2020.
Article in English | MEDLINE | ID: mdl-33193390

ABSTRACT

The suppressor of cytokine signaling (SOCS) family of intracellular checkpoint inhibitors has received little recognition compared to other checkpoint inhibitors. Two members of this family, SOCS1 and SOCS3, are indispensable, since SOCS1 knockout in mice results in neonatal death due to interferon gamma (IFNγ) induced inflammatory disease, and SOCS3 knockout leads to embryonic lethality. We have shown that SOCS1 and SOCS3 (SOCS1/3) function as virus induced intrinsic virulence factors for influenza A virus, EMC virus, herpes simplex virus 1 (HSV-1), and vaccinia virus infections. Other viruses such as pathogenic pig enteric coronavirus and coronavirus induced severe acute respiratory syndrome (SARS) spike protein also induce SOCS virus intrinsic virulence factors. SOCS1/3 exert their viral virulence effect via inhibition of type I and type II interferon (IFN) function. Specifically, the SOCS bind to the activation loop of receptor-associated tyrosine kinases JAK2 and TYK2 through the SOCS kinase inhibitory region (KIR), which inhibits STAT transcription factor activation by the kinases. Activated STATs are required for IFN function. We have developed a small peptide antagonist of SOCS1/3 that blocks SOCS1/3 inhibitory activity and prevents virus pathogenesis. The antagonist, pJAK2(1001-1013), is comprised of the JAK2 activation loop, phosphorylated at tyrosine 1007 with a palmitate for cell penetration. The remarkable thing about SOCS1/3 is that it serves as a broad, simple tool of perhaps most pathogenic viruses to avoid innate host IFN defense. We suggest in this Perspective that SOCS1/3 antagonist is a simple counter measure to SOCS1/3 and should be an effective mechanism as a prophylactic and/or therapeutic against the COVID-19 pandemic that is caused by coronavirus SARS-CoV2.


Subject(s)
COVID-19 Drug Treatment , COVID-19/immunology , SARS-CoV-2/physiology , Suppressor of Cytokine Signaling 1 Protein/immunology , Suppressor of Cytokine Signaling 3 Protein/immunology , Virulence Factors/immunology , Animals , COVID-19/genetics , COVID-19/virology , Humans , Interferons/genetics , Interferons/immunology , Mice , SARS-CoV-2/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Virulence Factors/genetics
7.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998457

ABSTRACT

MicroRNAs regulate gene expression of transcriptional factors, which influence Th17/Treg (regulatory T cells) balance, establishing the molecular mechanism of genetic and epigenetic regulation of Treg and Th17 cells is crucial for understanding rheumatoid arthritis (RA) pathogenesis. The study goal was to understand the potential impact of the selected microRNAs expression profiles on Treg/Th17 cells frequency, RA phenotype, the expression profile of selected microRNAs, and their correlation with the expression profiles of selected transcriptional factors: SOCS1, SMAD3, SMAD4, STAT3, STAT5 in RA; we used osteoarthritis (OA) and healthy controls (HCs) as controls. The study was conducted on 14 RA and 11 OA patients, and 15 HCs. Treg/Th17 frequency was established by flow cytometry. Gene expression analysis was estimated by qPCR. We noticed correlations in RA Th17 cells between miR-26 and SMAD3, STAT3, SOCS1; and miR-155 and STAT3-and in RA Treg cells between miR-26 and SOCS1; miR-31, -155 and SMAD3; and miR-155 and SMAD4. In RA Tregs, we found a negative correlation between miR-26, -126 and STAT5a. The expression level of miR-31 in Th17 cells from RA patients with DAS28 ≤ 5.1 is higher and that for miR-24 is greater in Tregs from patients with DAS28 > 5.1. MiR-146a in Tregs is higher in rheumatoid factor (RF) positive RA patients.


Subject(s)
Arthritis, Rheumatoid/genetics , MicroRNAs/genetics , Osteoarthritis/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Case-Control Studies , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Male , MicroRNAs/immunology , Middle Aged , Osteoarthritis/immunology , Osteoarthritis/pathology , Phenotype , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Severity of Illness Index , Smad3 Protein/genetics , Smad3 Protein/immunology , Smad4 Protein/genetics , Smad4 Protein/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
8.
Sci Rep ; 10(1): 15287, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943702

ABSTRACT

Th17/Treg imbalance contributes to chronic obstructive pulmonary disease (COPD) development and progression. However, intracellular signaling by suppressor of cytokine signaling (SOCS) 1 and SOCS3 and the proteins signal transducer and activator of transcription (STAT) 3 and STAT5 that orchestrate these imbalances are currently poorly understood. Thus, these proteins were investigated in C57BL/6 mice after exposure to cigarette smoke (CS) for 3 and 6 months. The expression of interleukin was measured by ELISA and the density of positive cells in peribronchovascular areas was quantified by immunohistochemistry. We showed that exposure to CS in the 3rd month first induced decreases in the numbers of STAT5+ and pSTAT5+ cells and the expression levels of TGF-ß and IL-10. The increases in the numbers of STAT3+ and pSTAT3+ cells and IL-17 expression occurred later (6th month). These findings corroborate the increases in the number of SOCS1+ cells in both the 3rd and 6th months, with concomitant decreases in SOCS3+ cells at the same time points. Our results demonstrated that beginning with the initiation of COPD development, there was a downregulation of the anti-inflammatory response mediated by SOCS and STAT proteins. These results highlight the importance of intracellular signaling in Th17/Treg imbalance and the identification of possible targets for future therapeutic approaches.


Subject(s)
Cytokines/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Disease Progression , Down-Regulation/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Suppressor of Cytokine Signaling 3 Protein/immunology
9.
Viruses ; 12(8)2020 07 25.
Article in English | MEDLINE | ID: mdl-32722523

ABSTRACT

Flaviviruses are constantly evolving diverse immune evasion strategies, and the exploitation of the functions of suppressors of cytokine signalling (SOCS) and protein inhibitors of activated STATs (PIAS) to favour virus replication has been described for Dengue and Japanese encephalitis viruses but not for yellow fever virus (YFV), which is still of global importance despite the existence of an effective vaccine. Some mechanisms that YFV employs to evade host immune defence has been reported, but the expression patterns of SOCS and PIAS in infected cells is yet to be determined. Here, we show that SOCS1 is down-regulated early in YFV-infected HeLa and HEK 293T cells, while SOCS3 and SOCS5 are not significantly altered, and PIAS mRNA expression appears to follow a rise-dip pattern akin to circadian-controlled genes. We also demonstrate that YFV evades interferon-ß application to produce comparable viral titres. This report provides initial insight into the in vitro expression dynamics of SOCS and PIAS upon YFV infection and a basis for further investigation into SOCS/PIAS expression and how these modulate the immune response in animal models.


Subject(s)
Down-Regulation , Suppressor of Cytokine Signaling 1 Protein/genetics , Virus Replication/immunology , Yellow fever virus/immunology , HEK293 Cells , HeLa Cells , Humans , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology
10.
Clin Exp Immunol ; 202(1): 47-59, 2020 10.
Article in English | MEDLINE | ID: mdl-32516488

ABSTRACT

Recurrence and diffuse infiltration challenge traditional therapeutic strategies for malignant glioma. Immunotherapy appears to be a promising approach to obtain long-term survival. Dendritic cells (DCs), the most specialized and potent antigen-presenting cells (APCs), play an important part in initiating and amplifying both the innate and adaptive immune responses against cancer cells. However, cancer cells can escape from immune surveillance by inhibiting maturation of DCs. Until the present, molecular mechanisms of maturation inhibition of DCs in the tumor microenvironment (TME) have not been fully revealed. Our study showed that pretreatment with tumor-conditioned medium (TCM) collected from supernatant of primary glioma cells significantly suppressed the maturation of DCs. TCM pretreatment significantly changed the morphology of DCs, TCM decreased the expression levels of CD80, CD83, CD86 and interleukin (IL)-12p70, while it increased the expression levels of IL-10, transforming growth factor (TGF)-ß and IL-6. RNA-Seq showed that TCM pretreatment significantly increased the gene expression level of suppressor of cytokine signaling 1 (SOCS1) in DCs. suppressor of cytokine signaling 1 (SOCS1) knock-down significantly antagonized the maturation inhibition of DCs by TCM, which was demonstrated by the restoration of maturation markers. TCM pretreatment also significantly suppressed T cell viability and T helper type 1 (Th1) response, and SOCS1 knock-down significantly antagonized this suppressive effect. Further, TCM pretreatment significantly suppressed p65 nuclear translocation and transcriptional activity in DCs, and SOCS1 knock-down significantly attenuated this suppressive effect. In conclusion, our research demonstrates that TCM up-regulate SOCS1 to suppress the maturation of DCs via the nuclear factor-kappa signaling pathway.


Subject(s)
Dendritic Cells/immunology , Glioma/immunology , NF-kappa B/immunology , Neoplasm Proteins/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Tumor Microenvironment/immunology , Dendritic Cells/pathology , Glioma/pathology , Humans , Th1 Cells/immunology , Th1 Cells/pathology , Tumor Cells, Cultured
11.
PLoS One ; 15(6): e0234479, 2020.
Article in English | MEDLINE | ID: mdl-32542025

ABSTRACT

There are differences in disease susceptibility to whirling disease (WD) among strains of rainbow trout. The North American strain Trout Lodge (TL) is highly susceptible, whereas the German Hofer (HO) strain is more resistant. The suppressor of cytokine signaling (SOCS) proteins are key in inhibiting cytokine signaling. Their role in modulating the immune response against whirling disease is not completely clear. This study aimed at investigating the transcriptional response of SOCS1 and SOCS3 genes to Myxobolus cerebralis along with that of several upstream regulators and immune response genes. M. cerebralis induced the expression of SOCS1, the IL-6-dependent SOCS3, the anti-inflammatory cytokine IL-10 and the Treg associated transcription factor FOXP3 in TL fish at multiple time points, which likely caused a restricted STAT1 and STAT3 activity affecting the Th17/Treg17 balance. The expression of SOCS1 and the IL-6-dependent SOCS3 was induced constraining the activation of STAT1 and STAT3 in TL fish, thereby causing Th17/Treg17 imbalance and leaving the fish unable to establish a protective immune response against M. cerebralis or control inflammatory reactions increasing susceptibility to WD. Conversely, in HO fish, the expression of SOCS1 and SOCS3 was restrained, whereas the expression of STAT1 and IL-23-mediated STAT3 was induced potentially enabling more controlled immune responses, accelerating parasite clearance and elevating resistance. The induced expression of STAT1 and IL-23-mediated STAT3 likely maintained a successful Th17/Treg17 balance and enabled fish to promote effective immune responses favouring resistance against WD. The results provide insights into the role of SOCS1 and SOCS3 in regulating the activation and magnitude of host immunity in rainbow trout, which may help us understand the mechanisms that underlie the variation in resistance to WD.


Subject(s)
Disease Susceptibility/immunology , Fish Diseases/immunology , Myxobolus/immunology , Oncorhynchus mykiss/immunology , Parasitic Diseases, Animal/immunology , STAT3 Transcription Factor/immunology , Suppressor of Cytokine Signaling 3 Protein/immunology , Animals , Oncorhynchus mykiss/parasitology , STAT1 Transcription Factor/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/cytology , Th17 Cells/immunology
12.
J Interferon Cytokine Res ; 40(4): 169-171, 2020 04.
Article in English | MEDLINE | ID: mdl-31985329

ABSTRACT

Both innate and adaptive immune responses of host are regulated by fine balance between negative and positive signals to ensure their termination and onset on entry of pathogens. Suppressor of cytokine signaling (SOCS) genes inhibit cytokine signaling pathways and regulate innate and adaptive immunity. SOCS genes perform their function by positive and negative regulation of macrophages, development and differentiation of T cells, and activation of dendritic cells. Although the role of SOCS1 and SOCS3 genes in hepatitis C virus (HCV) patients has been well studied, the correlation of SOCS1 gene expression with complete blood count in HCV-positive patients has not been established yet. We observed a weak positive correlation of SOCS1 gene expression with hemoglobin (Hb) and platelet count, whereas leukocytes were negatively correlated with the expression. This study also provides a comparative analysis of complete blood count between healthy subjects and HCV-positive subjects. The findings suggest that Hb and platelet count were significantly lower (P < 0.0001) in HCV patients than in healthy subjects.


Subject(s)
Blood Cells/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Adult , Cytokines/genetics , Cytokines/immunology , Female , Humans , Male , Suppressor of Cytokine Signaling 1 Protein/immunology
13.
Fish Shellfish Immunol ; 97: 146-152, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31846779

ABSTRACT

The suppressor of cytokine signaling (SOCS) was first described as inhibitors of cytokine signaling. The SOCS1, as a number of SOCS family, is an important negative regulator in the IFN signaling pathways in mammals. While data on functional characterization of SOCS1 in lower vertebrates are limited. In this study, we identified and characterized the full length SOCS1b gene of miiuy croaker (Miichthys miiuy). The sequence alignment analysis results showed that miiuy croaker SOCS1b (mmSOCS1b) have only a conserved SH2 domain that is similar to other vertebrates. To further study the functions of mmSOCS1b, we identified and determined its potential ability to perceive poly (I:C) stimulation. Stimulation experiments with poly (I:C) showed the significantly upregulated expression of mmSOCS1b in crucial immune-related tissues of spleen and kidney, indicating that mmSOCS1b might participate in the immune responses. Furthermore, the immunofluorescence assay indicated that mmSOCS1b present in the cytoplasmic of HeLa cells. In addition, mmSOCS1b could inhibit IFNα or IFNγ-induced ISRE reporter gene. In a word, we systematically and comprehensively analyzed the characterizations and functions of mmSOCS1b, which not only enriches the current knowledge of SOCS in IFN signaling regulation but also offer the basis for future research of fish SOCS family.


Subject(s)
Evolution, Molecular , Fish Proteins/genetics , Perciformes/genetics , Perciformes/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/immunology , Animals , Cytokines/immunology , Fish Proteins/immunology , HeLa Cells , Humans , Kidney/drug effects , Kidney/immunology , Phylogeny , Poly I-C/pharmacology , Sequence Alignment , Signal Transduction , Spleen/drug effects , Spleen/immunology
14.
Eur Rev Med Pharmacol Sci ; 23(18): 8068-8074, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31599432

ABSTRACT

OBJECTIVE: To clarify the effect of suppressor of cytokine signaling 1 (SOCS1) on diabetic nephropathy (DN)-induced renal injury by regulating the Toll-like receptor (TLR) signaling pathway. MATERIALS AND METHODS: The Sprague-Dawley rats were divided into control group (n=10) and DN group (established with streptozotocin injection, n=20). The DN rats were administrated with SOCS1 lentivirus to upregulate the in vivo expression. The rat blood glucose was detected to confirm the successful preparation of the DN model. The hepatic and renal function indexes, including blood urea nitrogen (BUN), alkaline phosphatase (ALP), alanine aminotransferase (ALT) and creatinine (CR) were detected. The pathological lesions in the kidney were observed via hematoxylin-eosin (HE) staining. Besides, the serum levels of the inflammatory factors in rats were detected via enzyme-linked immunosorbent assay (ELISA). The relative levels of genes in the TLR signaling pathway were detected via RT-PCR and Western blotting. RESULTS: The blood glucose level in rats of the DN group was significantly enhanced, indicating the successful modeling. The expression of SOCS1 was significantly upregulated in rats administrated with SOCS1 lentivirus. The contents of BUN, ALP, ALT, and CR in rats of SOCS1 overexpression group were significantly lower than those in the DN group. The inflammatory infiltration in the kidney and the glomerular injury were pronounced in the DN group. The serum levels of interleukin-1 (IL-1), interferon-γ (INF-γ), and tumor necrosis factor-α (TNF-α) were significantly declined in SOCS1 overexpression group. Besides, the mRNA expressions of myeloid differential protein-88 (MyD88), TLR2, and INF-γ, and the protein expression of TLR2 were all remarkably downregulated in SOCS1 overexpression group. CONCLUSIONS: SOCS1 can promote renal injury repair in DN rats by inhibiting the TLR pathway. Therefore, SOCS1 is expected to be a new target for the repair of DN renal injury.


Subject(s)
Diabetic Nephropathies/genetics , Interferon-gamma/genetics , Myeloid Differentiation Factor 88/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Toll-Like Receptor 2/genetics , Alanine Transaminase/metabolism , Alkaline Phosphatase/metabolism , Animals , Blood Glucose/metabolism , Blood Urea Nitrogen , Creatinine/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/immunology , Diabetic Nephropathies/pathology , Disease Models, Animal , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-1/metabolism , Kidney/pathology , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , RNA, Messenger/metabolism , Rats , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/immunology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
15.
Intervirology ; 62(2): 80-89, 2019.
Article in English | MEDLINE | ID: mdl-31315128

ABSTRACT

BACKGROUND: In immunocompromised patients, human cytomegalovirus (HCMV) infection is a major cause of morbidity and mortality. Suppressor of cytokine signaling (SOCS) proteins are very potent negative regulators of the janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways. We hypothesized that HCMV exploits SOCS1 and/or SOCS3 to its advantage. METHODS: All experiments were carried out with primary human lung-derived microvascular endothelial cells (HMVEC). SOCS1 and SOCS3 were silenced by transfecting the cells with siRNA. HCMV was propagated and titered on human lung-derived fibroblasts MRC5. Real-time PCR and Western blot were used to detect mRNA and protein levels, respectively. RESULTS: The data presented show that an efficient replication of HCMV in HMVEC is dependent on SOCS3 protein. Time course analysis revealed an increase in SOCS3 protein levels in infected cells. Silencing of SOCS3 (siSOCS3) resulted in inhibition of viral immediate early, early, and late antigen production. Consistently, HCMV titers produced by siSOCS3 cultures were significantly decreased when compared to control transfected cultures (siCNTRs). STAT1 and STAT2 phosphorylation was increased in siSOCS3-infected cells when compared to siCNTR-treated cells. CONCLUSION: These findings indicate the implication of SOCS3 in the mechanism of HCMV-mediated control of cellular immune responses.


Subject(s)
Cytomegalovirus/physiology , Endothelial Cells/virology , Immunity, Cellular , Suppressor of Cytokine Signaling 3 Protein/immunology , Virus Replication , Cells, Cultured , Endothelial Cells/immunology , Gene Silencing , Humans , Lung/cytology , Lung/virology , Phosphorylation , RNA, Small Interfering , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/immunology , Suppressor of Cytokine Signaling 3 Protein/genetics
16.
Nat Commun ; 9(1): 4964, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470758

ABSTRACT

The inflammasome plays a critical role in inflammation and immune responses against pathogens. However, whether or how inflammasome activation regulates type I interferon (IFN-I) signaling in the context of malaria infection remain unknown. Here we show mice deficient in inflammasome sensors AIM2, NLRP3 or adaptor Caspase-1 produce high levels of IFN-I cytokines and are resistant to lethal Plasmodium yoelii YM infection. Inactivation of inflammasome signaling reduces interleukin (IL)-1ß production, but increases IFN-I production. Mechanistically, we show inflammsome activation enhances IL-1ß-mediated MyD88-TRAF3-IRF3 signaling and SOCS1 upregulation. However, SOCS1 inhibits MyD88-IRF7-mediated-IFN-I signaling and cytokine production in plasmacytoid dendritic cells. By contrast, ablation of inflammsome components reduces SOCS1 induction, and relieves its inhibition on MyD88-IRF7-dependent-IFN-I signaling, leading to high levels of IFN-α/ß production and host survival. Our study identifies a previously unrecognized role of inflammasome activation in the negative regulation of IFN-I signaling pathways and provides potential targets for developing effective malaria vaccines.


Subject(s)
Inflammasomes/immunology , Interferon Regulatory Factor-7/immunology , Malaria/immunology , Myeloid Differentiation Factor 88/immunology , Plasmodium yoelii/physiology , Animals , Down-Regulation , Female , Humans , Inflammasomes/genetics , Interferon Regulatory Factor-7/genetics , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Malaria/genetics , Malaria/parasitology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology
17.
PLoS Pathog ; 14(8): e1007202, 2018 08.
Article in English | MEDLINE | ID: mdl-30075008

ABSTRACT

Gamma interferon (IFN-γ) is known to negatively regulate murine gammaherpesvirus-68 (MHV-68 or γHV-68) replication. This process involves the suppression of the viral gene replication and transcription activator (RTA) promoter, as well as activation of signal transducers and activators of transcription (STAT1). Notably, this effect is gradually attenuated during MHV-68 infection of bone marrow-derived macrophages (BMMs), which raised the possibility that the virus may utilize a mechanism that counteracts the antiviral effect of IFN-γ. By identifying the cellular factors that negatively regulate JAK-STAT1 signaling, we revealed that the infection of BMMs by MHV-68 induces the expression of suppressor of cytokine signaling 1 (SOCS1) and that depletion of SOCS1 restores the inhibitory effect of IFN-γ on virus replication. Moreover, we demonstrated that the expression of SOCS1 was induced as a result of the Toll-like receptor 3 (TLR3) mediated activation of the NF-κB signaling cascade. In conclusion, we report that TLR3-TRAF-NF-κB signaling pathway play a role in the induction of SOCS1 that counteracts the antiviral effect of IFN-γ during MHV-68 infection. This process is cell type-specific: it is functional in macrophages, but not in epithelial cells or fibroblasts. Our study reveals a mechanism that balances the immune responses and the escape of a gamma-herpesvirus in some antigen-presenting cells.


Subject(s)
Herpesviridae Infections/immunology , Interferon-gamma/metabolism , Macrophages/virology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Virus Replication/physiology , Animals , Herpesviridae Infections/metabolism , Host-Pathogen Interactions/immunology , Immune Evasion/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Rhadinovirus/physiology , Suppressor of Cytokine Signaling 1 Protein/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism
18.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29976680

ABSTRACT

AIDS-related human cytomegalovirus retinitis remains the leading cause of blindness among untreated HIV/AIDS patients worldwide. To study mechanisms of this disease, we used a clinically relevant animal model of murine cytomegalovirus (MCMV) retinitis with retrovirus-induced murine AIDS (MAIDS) that mimics the progression of AIDS in humans. We found in this model that MCMV infection significantly stimulates ocular suppressor of cytokine signaling 1 (SOCS1) and SOCS3, host proteins which hinder immune-related signaling by cytokines, including antiviral type I and type II interferons. The present study demonstrates that in the absence of retinal disease, systemic MCMV infection of mice without MAIDS, but not in mice with MAIDS, leads to mild stimulation of splenic SOCS1 mRNA. In sharp contrast, when MCMV is directly inoculated into the eyes of retinitis-susceptible MAIDS mice, high levels of intraocular SOCS1 and SOCS3 mRNA and protein are produced which are associated with significant intraocular upregulation of gamma interferon (IFN-γ) and interleukin-6 (IL-6) mRNA expression. We also show that infiltrating macrophages, granulocytes, and resident retinal cells are sources of intraocular SOCS1 and SOCS3 protein production during development of MAIDS-related MCMV retinitis, and SOCS1 and SOCS3 mRNA transcripts are detected in retinal areas histologically characteristic of MCMV retinitis. Furthermore, SOCS1 and SOCS3 are found in both MCMV-infected cells and uninfected cells, suggesting that these SOCS proteins are stimulated via a bystander mechanism during MCMV retinitis. Taken together, our findings suggest a role for MCMV-related stimulation of SOCS1 and SOCS3 in the progression of retinal disease during ocular, but not systemic, MCMV infection.IMPORTANCE Cytomegalovirus infection frequently causes blindness in untreated HIV/AIDS patients. This virus manipulates host cells to dysregulate immune functions and drive disease. Here, we use an animal model of this disease to demonstrate that cytomegalovirus infection within eyes during retinitis causes massive upregulation of immunosuppressive host proteins called SOCS. As viral overexpression of SOCS proteins exacerbates infection with other viruses, they may also enhance cytomegalovirus infection. Alternatively, the immunosuppressive effect of SOCS proteins may be protective against immunopathology during cytomegalovirus retinitis, and in such a case SOCS mimetics or overexpression treatment strategies might be used to combat this disease. The results of this work therefore provide crucial basic knowledge that contributes to our understanding of the mechanisms of AIDS-related cytomegalovirus retinitis and, together with future studies, may contribute to the development of novel therapeutic targets that could improve the treatment or management of this sight-threatening disease.


Subject(s)
Cytomegalovirus Retinitis/immunology , Immunosuppression Therapy , Murine Acquired Immunodeficiency Syndrome/immunology , Muromegalovirus/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Animals , Cytomegalovirus Retinitis/virology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Murine Acquired Immunodeficiency Syndrome/virology , Muromegalovirus/isolation & purification , Spleen/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Suppressor of Cytokine Signaling 3 Protein/immunology
19.
Mol Cancer Ther ; 17(9): 1941-1950, 2018 09.
Article in English | MEDLINE | ID: mdl-29891489

ABSTRACT

Ovarian cancer is the leading cause of gynecologic cancer-related deaths and novel therapeutic strategies are required. Programmed cell death 1 and programmed cell death ligand 1 (PD-L1), which are key mediators of host immune tolerance, are associated with ovarian cancer progression. Recent evidence indicates the importance of IFNγ-induced PD-L1 for immune tolerance in ovarian cancer. This study aimed to reveal the therapeutic potential of suppressor of cytokine signaling 1 (SOCS-1), an endogenous inhibitor of the Janus kinase (JAK)-STAT signaling pathway, for the treatment of ovarian cancer. IHC assessment revealed that patients with ovarian cancer with high intratumoral STAT1 activation exhibited poor prognosis compared with patients with low STAT1 activation (P < 0.05). Stimulation of OVISE, OVTOKO, OV2944-HM-1 (HM-1), and CT26 cell lines with IFNγ induced STAT1 phosphorylation and PD-L1 expression. Adenovirus-mediated SOCS-1 gene delivery (AdSOCS-1) in HM-1 and CT26 cells in vitro potently inhibited IFNγ-induced STAT1 phosphorylation and PD-L1 upregulation, similar to the addition of JAK inhibitor I, but failed to inhibit their proliferation. Notably, intratumoral injection of AdSOCS-1, but not AdLacZ, significantly inhibited the tumor growth of HM-1 and CT26 cells subcutaneously transplanted in immunocompetent syngeneic mice. AdSOCS-1 reduced PD-L1 expression on tumors and restored the activation of tumor-infiltrating CD8+ T cells. Moreover, the antitumor effect of AdSOCS-1 was significantly attenuated by PD-L1 Fc-fusion protein administration in vivo, suggesting that the effect of AdSOCS-1 is mainly attributable to enhancement of tumor immunity. This study highlights the potential clinical utility of SOCS-1 as an immune checkpoint inhibitor. Mol Cancer Ther; 17(9); 1941-50. ©2018 AACR.


Subject(s)
B7-H1 Antigen/immunology , Gene Expression Regulation, Neoplastic/immunology , Ovarian Neoplasms/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Female , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , T-Lymphocytes/metabolism , Transplantation, Homologous
20.
Clin Immunol ; 194: 9-18, 2018 09.
Article in English | MEDLINE | ID: mdl-29928998

ABSTRACT

Systemic juvenile idiopathic arthritis (sJIA) is a childhood rheumatic disease of unknown origin. Dysregulated innate immunity is implicated in disease pathology. We investigated if IL-1 inhibition affects circulating cytokines and monocyte gene expression. CD14+ monocytes from patients in the RAPPORT trial were analyzed by RT-PCR for expression of IL1B and transcription factors associated with monocyte activation. Serum IL-1ra decreased with treatment, and IL-18BP transiently increased. Serum levels of IL-1ß, IL-6, IL-10 and IL-18 were unchanged. IRF5 and STAT6 were decreased, and PPARG was increased, independent of clinical response, and may represent a skew toward a PPARG-driven M2-like phenotype. IL1B expression was decreased in early clinical responders. A transient increase in STAT1, and a decrease in SOCS1 preceded the reduction in IL1B in early clinical responders. Changes in IL1B/STAT1/SOCS1 could be associated with crosstalk between IL-1 and IFN pathways in sJIA. These transcriptional changes might be useful as drug response biomarkers.


Subject(s)
Arthritis, Juvenile/drug therapy , Interleukin-1/antagonists & inhibitors , Monocytes/drug effects , Recombinant Fusion Proteins/therapeutic use , Arthritis, Juvenile/immunology , Clinical Trials as Topic , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Interleukin-1/immunology , Interleukin-1beta/immunology , Monocytes/immunology , Randomized Controlled Trials as Topic , STAT1 Transcription Factor/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...